Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Cancer Cell Int ; 22(1): 153, 2022 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-35436935

RESUMO

BACKGROUND: Growing evidence has indicated that tumor-associated macrophages (TAMs) promote tumor angiogenesis. However, the mechanisms underlying the pro-angiogenic switch of TAMs remains unclear. Here, we examined how exosomal miR-301a-3p secreted by esophageal squamous cell carcinoma (ESCC) cells triggers the pro-angiogenic switch of TAMs. METHODS: We quantified miR-301a-3p levels in ESCC tumors using qRT-PCR. Macrophage phenotypes were identified using flow cytometry and qRT-PCR. The pro-angiogenic ability of TAMs was measured using the CCK-8 assay, scratch assay, Transwell migration and invasion assay, and tube formation assay. The mechanism by which exosomal miR-301a-3p secreted by ESCC cells triggers the pro-angiogenic switch of TAMs was elucidated using western blots, qRT-PCR, and a dual-luciferase reporter assay. RESULTS: We observed anomalous miR-301a-3p overexpression in ESCC tumor tissues and cell lines. Then, we verified that ESCC-derived exosomes promoted angiogenesis by inducing macrophage polarization into M2 type, and exosomal miR-301a-3p secreted by ESCC cells was responsible for this effect. Finally, we discovered that exosomal miR-301a-3p promoted M2 macrophage polarization via the inhibition of PTEN and activation of the PI3K/AKT signaling pathway, subsequently promoting angiogenesis via the secretion of VEGFA and MMP9. CONCLUSION: The pro-angiogenic switch of TAMs is triggered by exosomal miR-301a-3p secreted from ESCC cells via the PTEN/PI3K/AKT signaling pathway. Although tumor angiogenesis can be regulated by a wide range of factors, exosomal miR-301a-3p could hold promise as a novel anti-angiogenesis target for ESCC treatment.

2.
J Carcinog ; 20: 18, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34729050

RESUMO

BACKGROUND: The TP53 tumor suppressor gene is the most commonly mutated gene in human cancers. Humans who inherit mutant TP53 alleles develop a wide range of early onset cancers, a disorder called Li-Fraumeni Syndrome (LFS). Trp53-deficient mice recapitulate most but not all of the cancer phenotypes observed in TP53-deficient human cancers, indicating that new animal models may complement current mouse models and better inform on human disease development. MATERIALS AND METHODS: The recent application of CRISPR/Cas9 genetic engineering technology has permitted the emergence of golden Syrian hamsters as genetic models for wide range of diseases, including cancer. Here, the first cancer phenotype of TP53 knockout golden Syrian hamsters is described. RESULTS: Hamsters that are homozygous for TP53 mutations become moribund on average ~ 139 days of age, while hamsters that are heterozygous become moribund at ~ 286 days. TP53 homozygous knockout hamsters develop a wide range of cancers, often synchronous and metastatic to multiple tissues, including lymphomas, several sarcomas, especially hemangiosarcomas, myeloid leukemias and several carcinomas. TP53 heterozygous mutants develop a more restricted tumor spectrum, primarily lymphomas. CONCLUSIONS: Overall, hamsters may provide insights into how TP53 deficiency leads to cancer in humans and can become a new model to test novel therapies.

3.
J Cell Physiol ; 234(10): 17775-17785, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30864145

RESUMO

Circular RNAs have been found to be aberrantly expressed in tumors and their significance in tumorigenesis has been focused on. The role of circDYNC1H1 in hepatocellular carcinoma (HCC) pathogenesis and its relationship with miR-140-5p were explored. The expression of circDYNC1H1, miR-140-5p, and SULT2B1 in HCC tissues and cells was measured, and Pearson's analysis was used to analyze their expression correlation. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Transwell assays were performed to determine cell proliferation and migration. Binding between circDYNC1H1 and miR-140-5p was evaluated with RNA pull-down assay. A luciferase reporter assay was conducted to assess the interaction between circDYNC1H1 and miR-140-5p and between miR-140-5p and SULT2B1. circDYNC1H1 was highly expressed in HCC tissues (n = 20), and it was negatively associated with the expression of miR-140-5p but positively correlated with SULT2B1 messenger RNA expression. circDYNC1H1 was upregulated in cell lines of HCC. Interference of circDYNC1H1 suppressed cell proliferation and migration of HCC. circDYNC1H1 acted as a sponge of miR-140-5p. miR-140-5p controlled SULT2B1 expression by targeting its 3'-untranslated region. circDYNC1H1 enhanced SULT2B1 expression via sponging miR-140-5p. Downregulation of circDYNC1H1 disturbed cell proliferation and migration of HCC through miR-140-5p/SULT2B1 pathway. Silencing of circDYNC1H1 delayed tumor growth in HCC mouse model. Acting like a sponge of miR-140-5p, silenced circDYNC1H1 downregulated SULT2B1 to restrain HCC cell proliferation and migration, which is adverse to HCC growth and progression.


Assuntos
Carcinoma Hepatocelular/genética , Movimento Celular/genética , Proliferação de Células/genética , Dineínas do Citoplasma/genética , Regulação para Baixo/genética , Neoplasias Hepáticas/genética , MicroRNAs/genética , Regiões 3' não Traduzidas/genética , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Regulação para Cima/genética
4.
Cell Physiol Biochem ; 41(5): 1981-1992, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28478444

RESUMO

BACKGROUND: MiR-34a is identified as a tumor suppressor gene and involved in acute myeloid leukemia (AML) development. However, the regulatory mechanism of miR-34a in AML is unclear. METHODS: The expression of miR-34a and HMGB1 in HL-60, THP-1 and HS-5 cells were detected by qRT-PCR and western blot. Lipofectamine 2000 was used to transfect with miR-34a mimics, miR-34a inhibitor, si-HMGB1, pcDNA 3.1-HMGB1, and corresponding controls. The apoptosis and autophagy of transfected AML cells were assessed by flow cytometry and western blot, respectively. Bioinformatics software and dual luciferase reporter assay were applied to predict and verify the target of miR-34a. The effects of miR-34a mimics or si-HMGB1 on chemotherapy-induced autophagy were further explored in HL-60 cells treated with all-trans retinoic acid (ATRA) along with lysosomal protease inhibitors E64d and pepstatin A. RESULTS: MiR-34a was lower expressed and HMGB1 mRNA and proteins were both higher expressed in HL-60 and THP-1 cells compared with that in HS-5 cells. Higher expression levels of MiR-34 and lower expression levels of HMGB1 both significantly promoted apoptosis and inhibited autophagy in HL-60 and THP-1 cells. Dual luciferase reporter system confirmed that HMGB1 was a potential target of miR-34a. Moreover, overexpression of HMGB1 dramatically reversed the promotion of apoptosis and inhibition of autophagy mediated by higher expression level of miR-34a. Higher expression level of miR-34a and lower expression level of HMGB1 both inhibited chemotherapy-induced autophagy by stimulating the LC3 conversion. CONCLUSION: MiR-34a promoted cell apoptosis and inhibited autophagy by targeting HMGB1. Therefore, miR-34a may be a potential promising molecular target for AML therapy.


Assuntos
Apoptose , Autofagia , Proteína HMGB1/biossíntese , Leucemia Mieloide Aguda/metabolismo , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/biossíntese , Células HL-60 , Proteína HMGB1/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Neoplásico/genética
5.
AJR Am J Roentgenol ; 209(5): 1033-1038, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28871809

RESUMO

OBJECTIVE: The purpose of this study was to use virtual monochromatic spectral CT to investigate the usefulness of iodine concentration (IC) and its correlation with clinicopathologically determined prognostic factors in gastric adenocarcinoma. SUBJECTS AND METHODS: From June 2012 to March 2015, 34 patients with gastric adenocarcinoma underwent arterial and portal venous phase spectral CT. The ICs in the arterial and portal venous phases were calculated and then normalized with the aorta as normalized IC (NIC). The surgical specimen was evaluated with CD34 staining to determine microvessel density (MVD). The correlation between imaging results and clinicopathologic findings was investigated for histologic grading, lymph node metastasis, serosal involvement, distant metastasis, pathologic TNM stage, and MVD. RESULTS: The mean arterial phase NIC value of tumors was 0.12 ± 0.03, portal venous phase NIC value was 0.39 ± 0.06, and MVD was 26.94 ± 7.87 vessels per high-power field (×400). Both arterial phase and portal venous phase NIC values were significantly higher in poorly differentiated gastric adenocarcinomas (p = 0.005) than in moderately differentiated tumors (p = 0.013). There was no significant correlation between NIC and serosal involvement or distant metastasis. There was significant correlation between the NIC and MVD in gastric adenocarcinoma (arterial phase NIC, p = 0.013; portal venous phase NIC, p = 0.001). However, neither the arterial nor the portal venous phase NIC of gastric adenocarcinoma had a significant relation to lymphatic metastasis or pathologic TNM stage. There was a significant difference between the high and low MVD groups with respect to portal venous phase NIC (p = 0.045). CONCLUSION: NIC can serve as a useful predictor of angiogenesis and degree of differentiation of moderately and poorly differentiated gastric adenocarcinomas.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/patologia , Iodo/metabolismo , Neoplasias Gástricas/diagnóstico por imagem , Neoplasias Gástricas/patologia , Tomografia Computadorizada por Raios X , Adenocarcinoma/metabolismo , Idoso , Meios de Contraste , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Neoplasias Gástricas/metabolismo , Ácidos Tri-Iodobenzoicos
6.
Pak J Pharm Sci ; 27(4 Suppl): 1063-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25016267

RESUMO

This paper is to study the effect of traditional Chinese medicine monomer including quercetin, curcumin and Glaucocalyxin A on Hela cell of cervical cancer. The inhibiting effect of quercetin, curcumin and Glaucocalyxin A on HeLa cells' proliferation is detected through using MTT method. Analysis for the effect of quercetin, curcumin and Glaucocalyxin A on proliferation cycle of Hela cell is performed through adopting flow cytometry. Three kinds of traditional Chinese medicine monomer can inhibit the growth of Hela cell, and they show dependent relationship between time and dose. Quercetin, curcumin and Glaucocalyxin A could inhibit cell proliferation, probably through making Hela cell be in stagnation and inducing its apoptosis.


Assuntos
Curcumina/farmacologia , Diterpenos do Tipo Caurano/farmacologia , Medicina Tradicional Chinesa , Quercetina/farmacologia , Proliferação de Células/efeitos dos fármacos , Células HeLa , Humanos
7.
Pak J Pharm Sci ; 27(5 Suppl): 1651-6, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25262513

RESUMO

The clinical study found that NEDD9 showed high expression on the invasion in gastric cancer tissues and metastasis of the tumor. Based on promoting the fundamental role (Sisen et al., 2013) to the expression level, the author further study NEDD9 siRNA, which could significantly reduce NEDD9 protein and mRNA in gastric cancer BGC823 cells, inhibition of cell proliferation, induce cell apoptosis, and decrease the invasiveness of gastric cancer cells, suggesting that NEDD9 plays an important role in the gastric cancer cell proliferation, apoptosis and invasion force. Through constructing a model transplanted gastric cancer in nude mice, the author observes the effect of NEDD9 siRNA on the growth of gastric cancer x-engrafts, and application of NEDD9 immunohistochemical SP method. The author also uses Western blot method to detect the gastric carcinoma in nude mice transplanted tumor tissues expression; applies situ hybridization, RT-PCR technology to detect the gastric cancer engraft tissues in NEDD9 mRNA. In order to further explore the relationship between NEDD9 and the development of gastric cancer, he provides a theoretical basis for the NEDD9 targeted therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , RNA Interferente Pequeno/genética , Neoplasias Gástricas/patologia , Animais , Xenoenxertos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , RNA Mensageiro/análise , Neoplasias Gástricas/genética
8.
Pak J Pharm Sci ; 27(5 Suppl): 1675-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25262517

RESUMO

The research protocol is MTT (Methyl Thiazolyl Tetrazolium) method, Hoechst33342 staining method and flow cytometry detection to observe the effect of minor bupleurum decoction on proliferation inhibition and apoptosis-inducing of esophageal cancer cell strain Eca-109 cell and its purpose is to discuss the effect. The result of MTT method shows that minor buplerum decoction can obviously inhibit proliferation of esophageal cancer cell strain Eca-109 cell. Apoptosis number of esophageal cancer cell increased with the increase of concentration of tetrandrine by the Hoechst 35528 staining experiment of cancer cell in three different concentrations. Flow cytometry detection result showed that cells in cell cycle G0/G1 of esophageal cancer cell strain Eca-109 cell increased obviously and cell in s period decreased significantly. This research proved that minor bupleurum decoction had anti-tumor effect and can influent proliferation and apoptosis of esophageal cancer cell strain Eca-109 cell.


Assuntos
Apoptose/efeitos dos fármacos , Bupleurum , Proliferação de Células/efeitos dos fármacos , Neoplasias Esofágicas/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Esofágicas/patologia , Humanos
9.
Clin Epigenetics ; 16(1): 25, 2024 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-38336771

RESUMO

RATIONALE: Cancer of unknown primary (CUP) is a group of rare malignancies with poor prognosis and unidentifiable tissue-of-origin. Distinct DNA methylation patterns in different tissues and cancer types enable the identification of the tissue of origin in CUP patients, which could help risk assessment and guide site-directed therapy. METHODS: Using genome-wide DNA methylation profile datasets from The Cancer Genome Atlas (TCGA) and machine learning methods, we developed a 200-CpG methylation feature classifier for CUP tissue of origin prediction (MFCUP). MFCUP was further validated with public-available methylation array data of 2977 specimens and targeted methylation sequencing of 78 Formalin-fixed paraffin-embedded (FFPE) samples from a single center. RESULTS: MFCUP achieved an accuracy of 97.2% in a validation cohort (n = 5923) representing 25 cancer types. When applied to an Infinium 450 K array dataset (n = 1052) and an Infinium EPIC (850 K) array dataset (n = 1925), MFCUP achieved an overall accuracy of 93.4% and 84.8%, respectively. Based on MFCUP, we established a targeted bisulfite sequencing panel and validated it with FFPE sections from 78 patients of 20 cancer types. This methylation sequencing panel correctly identified tissue of origin in 88.5% (69/78) of samples. We also found that the methylation levels of specific CpGs can distinguish one cancer type from others, indicating their potential as biomarkers for cancer diagnosis and screening. CONCLUSION: Our methylation-based cancer classifier and targeted methylation sequencing panel can predict tissue of origin in diverse cancer types with high accuracy.


Assuntos
Metilação de DNA , Neoplasias Primárias Desconhecidas , Humanos , Neoplasias Primárias Desconhecidas/diagnóstico , Neoplasias Primárias Desconhecidas/genética , Análise de Sequência de DNA
10.
Clin Exp Med ; 24(1): 42, 2024 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-38400850

RESUMO

Infection is the leading cause of morbidity and mortality in patients with multiple myeloma (MM). Studying the relationship between different traits of Coronavirus 2019 (COVID-19) and MM is critical for the management and treatment of MM patients with COVID-19. But all the studies on the relationship so far were observational and the results were also contradictory. Using the latest publicly available COVID-19 genome-wide association studies (GWAS) data, we performed a bidirectional Mendelian randomization (MR) analysis of the causality between MM and different traits of COVID-19 (SARS-CoV-2 infection, COVID-19 hospitalization, and severe COVID-19) and use multi-trait analysis of GWAS(MTAG) to identify new associated SNPs in MM. We performed co-localization analysis to reveal potential causal pathways between diseases and over-representation enrichment analysis to find involved biological pathways. IVW results showed SARS-CoV-2 infection and COVID-19 hospitalization increased risk of MM. In the reverse analysis, the causal relationship was not found between MM for each of the different symptoms of COVID-19. Co-localization analysis identified LZTFL1, MUC4, OAS1, HLA-C, SLC22A31, FDX2, and MAPT as genes involved in COVID-19-mediated causation of MM. These genes were mainly related to immune function, glycosylation modifications and virus defense. Three novel MM-related SNPs were found through MTAG, which may regulate the expression of B3GNT6. This is the first study to use MR to explore the causality between different traits of COVID-19 and MM. The results of our two-way MR analysis found that SARS-CoV-2 infection and COVID-19 hospitalization increased the susceptibility of MM.


Assuntos
COVID-19 , Mieloma Múltiplo , Humanos , Teorema de Bayes , Estudo de Associação Genômica Ampla , Análise da Randomização Mendeliana , Mieloma Múltiplo/genética , COVID-19/genética , SARS-CoV-2/genética
11.
Transl Cancer Res ; 13(3): 1367-1381, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38617526

RESUMO

Background: Head and neck squamous cell carcinoma (HNSCC) is the most common type and accounts for 90% of all head and neck cancer cases. Despite advances in early diagnosis and treatment strategies-chemotherapy, surgical resection, and radiotherapy-5-year survival remains grim. For patients with early-stage HNSCC, accurately predicting clinical outcomes is challenging. Considering the pivotal role of the immune system in HNSCC, we developed a reliable immune-related gene signature (IRGS) and explored its predictive accuracy in patients with early-stage HNSCC. Methods: We examined immune gene expression profiles and clinical information from 230 early-stage HNSCC specimens, including 100 cases from The Cancer Genome Atlas (TCGA), 49 cases from the Gene Expression Omnibus (GEO; GSE65858), and 81 cases from an independent clinical cohort. The prognostic signature was constructed using Kaplan-Meier analysis and the least absolute shrinkage and selection operator (LASSO) Cox algorithm. We also explored the IRGS-related biological pathways and immune landscape using bioinformatics analysis. Results: A nine-immune-gene signature was generated to significantly stratify patients into high and low-risk groups. High risk patients exhibited shorter survival time [hazard ratio (HR) =13.795, 95% confidence interval (CI): 3.275-58.109, P<0.001]. The signature demonstrated robust prognostic ability in the training and validation sets and could independently predict overall survival (OS) and relapse-free survival (RFS). Subsequently, the receiver operating characteristic (ROC) curve and C-index confirmed the signature's predictive accuracy compared to clinical parameters. Additionally, cases classified as low risk showed more immune cell infiltration than high-risk cases. Conclusions: Our novel IRGS is a reliable and robust classifier for accurate patient stratification and prognostic evaluation. Future studies will attempt to affirm the signature's clinical application to early-stage HNSCC.

12.
Comput Biol Med ; 178: 108710, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38843570

RESUMO

BACKGROUND: Efficient and precise diagnosis of non-small cell lung cancer (NSCLC) is quite critical for subsequent targeted therapy and immunotherapy. Since the advent of whole slide images (WSIs), the transition from traditional histopathology to digital pathology has aroused the application of convolutional neural networks (CNNs) in histopathological recognition and diagnosis. HookNet can make full use of macroscopic and microscopic information for pathological diagnosis, but it cannot integrate other excellent CNN structures. The new version of HookEfficientNet is based on a combination of HookNet structure and EfficientNet that performs well in the recognition of general objects. Here, a high-precision artificial intelligence-guided histopathological recognition system was established by HookEfficientNet to provide a basis for the intelligent differential diagnosis of NSCLC. METHODS: A total of 216 WSIs of lung adenocarcinoma (LUAD) and 192 WSIs of lung squamous cell carcinoma (LUSC) were recruited from the First Affiliated Hospital of Zhengzhou University. Deep learning methods based on HookEfficientNet, HookNet and EfficientNet B4-B6 were developed and compared with each other using area under the curve (AUC) and the Youden index. Temperature scaling was used to calibrate the heatmap and highlight the cancer region of interest. Four pathologists of different levels blindly reviewed 108 WSIs of LUAD and LUSC, and the diagnostic results were compared with the various deep learning models. RESULTS: The HookEfficientNet model outperformed HookNet and EfficientNet B4-B6. After temperature scaling, the HookEfficientNet model achieved AUCs of 0.973, 0.980, and 0.989 and Youden index values of 0.863, 0.899, and 0.922 for LUAD, LUSC and normal lung tissue, respectively, in the testing set. The accuracy of the model was better than the average accuracy from experienced pathologists, and the model was superior to pathologists in the diagnosis of LUSC. CONCLUSIONS: HookEfficientNet can effectively recognize LUAD and LUSC with performance superior to that of senior pathologists, especially for LUSC. The model has great potential to facilitate the application of deep learning-assisted histopathological diagnosis for LUAD and LUSC in the future.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Aprendizado Profundo , Neoplasias Pulmonares , Redes Neurais de Computação , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Interpretação de Imagem Assistida por Computador/métodos , Diagnóstico por Computador/métodos
13.
Neuromolecular Med ; 26(1): 29, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39014255

RESUMO

Vascular dementia (VaD) is a cognitive disorder characterized by a decline in cognitive function resulting from cerebrovascular disease. The hippocampus is particularly susceptible to ischemic insults, leading to memory deficits in VaD. Astaxanthin (AST) has shown potential therapeutic effects in neurodegenerative diseases. However, the mechanisms underlying its protective effects in VaD and against hippocampal neuronal death remain unclear. In this study, We used the bilateral common carotid artery occlusion (BCCAO) method to establish a chronic cerebral hypoperfusion (CCH) rat model of VaD and administered a gastric infusion of AST at 25 mg/kg per day for 4 weeks to explore its therapeutic effects. Memory impairments were assessed using Y-maze and Morris water maze tests. We also performed biochemical analyses to evaluate levels of hippocampal neuronal death and apoptosis-related proteins, as well as the impact of astaxanthin on the PI3K/Akt/mTOR pathway and oxidative stress. Our results demonstrated that AST significantly rescued memory impairments in VaD rats. Furthermore, astaxanthin treatment protected against hippocampal neuronal death and attenuated apoptosis. We also observed that AST modulated the PI3K/Akt/mTOR pathway, suggesting its involvement in promoting neuronal survival and synaptic plasticity. Additionally, AST exhibited antioxidant properties, mitigating oxidative stress in the hippocampus. These findings provide valuable insights into the potential therapeutic effects of AST in VaD. By elucidating the mechanisms underlying the actions of AST, this study highlights the importance of protecting hippocampal neurons and suggests potential targets for intervention in VaD. There are still some unanswered questions include long-term effects and optimal dosage of the use in human. Further research is warranted to fully understand the therapeutic potential of AST and its application in the clinical treatment of VaD.


Assuntos
Apoptose , Demência Vascular , Hipocampo , Transtornos da Memória , Neurônios , Fármacos Neuroprotetores , Estresse Oxidativo , Ratos Sprague-Dawley , Xantofilas , Animais , Xantofilas/uso terapêutico , Xantofilas/farmacologia , Hipocampo/efeitos dos fármacos , Demência Vascular/tratamento farmacológico , Ratos , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Estresse Oxidativo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Modelos Animais de Doenças , Transdução de Sinais/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Morte Celular/efeitos dos fármacos , Antioxidantes/uso terapêutico , Antioxidantes/farmacologia , Teste do Labirinto Aquático de Morris/efeitos dos fármacos
14.
Pathol Oncol Res ; 29: 1611081, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37168048

RESUMO

Despite significant advances in the diagnosis and treatment of esophageal squamous cell carcinoma (ESCC), esophageal cancer is still a heavy social and medical burden due to its high incidence. Uncontrolled division and proliferation is one of the characteristics of tumor cells, which will promote rapid tumor growth and metastasis. Early mitotic inhibitor 1 (Emi1), ubiquitin-conjugating enzyme 10 (UBCH10) and CyclinB1 are important proteins involved in the regulation of cell cycle. In this study, the expression of Emi1, UBCH10 and CyclinB1 in ESCC tissues and adjacent normal tissues will be analyzed by immunohistochemistry and in-situ hybridization techniques, and their relationship with tumor proliferation and apoptosis will be analyzed. The results showed that Emi1, UBCH10 and CyclinB1 genes and proteins were highly expressed in tumor tissues, which were correlated with tumor grade, lymph node metastasis and pathological stage, and positively correlated with tumor proliferation. Emi1, UBCH10 and CyclinB1 are also positively correlated. It is speculated that Emi1, UBCH10 and CyclinB1 genes synergically promote tumor proliferation and inhibit apoptosis, which may be potential diagnostic and therapeutic targets for ESCC.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Humanos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Regulação Neoplásica da Expressão Gênica , Imuno-Histoquímica , Enzimas de Conjugação de Ubiquitina/genética
15.
Int J Oncol ; 62(6)2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37144499

RESUMO

Multiple myeloma (MM) is one of the three major malignancies of the hematological system in middle­aged and older individuals. The incidence of MM increases with age and due to its drug resistance and high recurrence, MM seriously harms human health. Long non­coding RNAs (lncRNAs) are RNA molecules with a length of >200 nt and rarely encode proteins. Numerous studies reported that lncRNAs regulate carcinogenesis and cancer progression. MM­associated lncRNAs affect features of tumor cells, including proliferation, apoptosis, adhesion and treatment resistance. The present review aims to summarize the latest findings on the roles of lncRNAs in MM to deepen the understanding of this field and provide insight for developing specific diagnostic tools and effective treatment strategies for MM, including novel biomarkers and targeted lncRNA therapeutics.


Assuntos
Mieloma Múltiplo , RNA Longo não Codificante , Humanos , Pessoa de Meia-Idade , Idoso , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica , Carcinogênese/genética
16.
Int Immunopharmacol ; 117: 109827, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36989973

RESUMO

BACKGROUND: Therapeutic options for small cell lung cancer (SCLC), a particularly lethal malignancy, remain limited. Members of the B7-CD28 family are compelling targets for immune checkpoint blockade strategies, which involve activating, inhibiting, and fine-tuning the T cell immune response. However, their clinical features and significance have not been explored comprehensively. METHOD: We enrolled 228 patients with an initial diagnosis of SCLC, including 77 cases from Cbioportal and a validation cohort of 151 cases with qPCR data. Kaplan-Meier analysis and LASSO Cox model were used to identify a signature based on the B7-CD28 family, which was applied for accurate prediction of chemotherapy benefit and prognosis for SCLC patients. In addition, we applied bioinformatics analysis to explore potential signature-related molecular mechanisms and the immune landscape. RESULTS: The mutation profiles of healthy tissues and SCLC tissues were distinct. A signature consisting of seven genes (CD86, ICOSLG, CD276, CD28, CTLA-4, PDCD1, and TMIGD2) was identified and applied to group patients based on risk level (high-risk and low-risk), producing two groups for which survival outcomes differed significantly (HR = 3.81, 95% CI: 2.16-6.74, P < 0.001). The immune checkpoint-based signature accurately predicted patient outcomes for the selected training and validation sets. Notably, low-risk patients were more likely to benefit from chemotherapy and showed greater immune activation. Additionally, time-dependent ROC curves and C-index analysis confirmed that the immune checkpoint-based signature has excellent predictive power for prognosis and chemotherapy benefit compared to clinically recognized parameters. Finally, multivariate analysis confirmed the identified signature as an independent risk factor for prognosis and chemotherapeutic response. CONCLUSION: We systematically obtained a comprehensive molecular profile for B7-CD28 family members in SCLC patients, from which we produced a reliable and robust prognostic immune checkpoint-based signature with the potential to improve prognostic stratification and therapy strategies for SCLC patients.


Assuntos
Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/genética , Antígenos CD28/genética , Prognóstico , Fatores de Transcrição , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Antígenos B7
17.
Cancer Med ; 12(13): 14468-14483, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37184125

RESUMO

BACKGROUND: Esophageal squamous cell carcinoma (ESCC), an aggressive gastrointestinal tumor, often has high early lymphatic metastatic potential. Cancer-associated fibroblasts (CAFs) are primary components in tumor microenvironment (TME), and the impact of CAFs and its derived exosomes on lymphangiogenesis remains elusive. MATERIALS AND METHODS: CAFs and the microlymphatic vessel density (MLVD) in ESCC was examined. Exosomes were extracted from primary normal fibroblast (NFs) and CAFs. Subsequently, tumor-associated lymphatic endothelial cells (TLECs) were treated with these exosomes, and the effect on their biological behavior was examined. miR-100-5p was selected as the target miRNA, and its effect on TLECs was examined. The target of miR-100-5p was predicted and confirmed. Subsequently, IGF1R, PI3K, AKT, and p-AKT expression in TLECs and tumors treated with exosomes and miR-100-5p were examined. RESULTS: A large number of CAFs and microlymphatic vessels were present in ESCC, leading to a poor prognosis. CAF-derived exosomes promoted proliferation, migration, invasion, and tube formation in TLECs. Further, they also enhanced lymphangiogenesis in ESCC xenografts. miR-100-5p levels were significantly lower in CAF-derived exosomes than in NF-derived exosomes. miR-100-5p inhibited proliferation, migration, invasion, and tube formation in TLECs. Further, miR-100-5p inhibited lymphangiogenesis in ESCC xenografts. Mechanistic studies revealed that this inhibition was mediated by the miR-100-5p-induced inhibition of IGF1R/PI3K/AKT axis. CONCLUSION: Taken together, our study demonstrates that CAF-derived exosomes with decreased miR-100-5p levels exhibit pro-lymphangiogenesis capacity, suggesting a possibility of targeting IGF1R/PI3K/AKT axis as a strategy to inhibit lymphatic metastasis in ESCC.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Exossomos , MicroRNAs , Humanos , Carcinoma de Células Escamosas do Esôfago/patologia , Neoplasias Esofágicas/patologia , Regulação para Baixo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Exossomos/metabolismo , Linfangiogênese/genética , Células Endoteliais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proliferação de Células , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo , Regulação Neoplásica da Expressão Gênica , Movimento Celular , Microambiente Tumoral/genética
18.
EBioMedicine ; 92: 104587, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37149929

RESUMO

BACKGROUND: Metastasis is one of the most lethal hallmarks of esophageal squamous cell carcinoma (ESCC), yet the mechanisms remain unclear due to a lack of reliable experimental models and systematic identification of key drivers. There is urgent need to develop useful therapies for this lethal disease. METHODS: A genome-wide CRISPR/Cas9 screening, in combination with gene profiling of highly invasive and metastatic ESCC sublines, as well as PDX models, was performed to identify key regulators of cancer metastasis. The Gain- and loss-of-function experiments were taken to examine gene function. Protein interactome, RNA-seq, and whole genome methylation sequencing were used to investigate gene regulation and molecular mechanisms. Clinical significance was analyzed in tumor tissue microarray and TCGA databases. Homology modeling, modified ELISA, surface plasmon resonance and functional assays were performed to identify lead compound which targets MEST to suppress cancer metastasis. FINDINGS: High MEST expression was associated with poor patient survival and promoted cancer invasion and metastasis in ESCC. Mechanistically, MEST activates SRCIN1/RASAL1-ERK-snail signaling by interacting with PURA. miR-449a was identified as a direct regulator of MEST, and hypermethylation of its promoter led to MEST upregulation, whereas systemically delivered miR-449a mimic could suppress tumor metastasis without overt toxicity. Furthermore, molecular docking and computational screening in a small-molecule library of 1,500,000 compounds and functional assays showed that G699-0288 targets the MEST-PURA interaction and significantly inhibits cancer metastasis. INTERPRETATION: We identified the MEST-PURA-SRCIN1/RASAL1-ERK-snail signaling cascade as an important mechanism underlying cancer metastasis. Blockade of MEST-PURA interaction has therapeutic potential in management of cancer metastasis. FUNDING: This work was supported by National Key Research and Development Program of China (2021YFC2501000, 2021YFC2501900, 2017YFA0505100); National Natural Science Foundation of China (31961160727, 82073196, 81973339, 81803551); NSFC/RGC Joint Research Scheme (N_HKU727/19); Natural Science Foundation of Guangdong Province (2021A1515011158, 2021A0505030035); Key Laboratory of Guangdong Higher Education Institutes of China (2021KSYS009).


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , MicroRNAs , Humanos , Carcinoma de Células Escamosas do Esôfago/patologia , Neoplasias Esofágicas/genética , Simulação de Acoplamento Molecular , Sistemas CRISPR-Cas , Detecção Precoce de Câncer , MicroRNAs/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células , Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição/genética
19.
Cell Discov ; 9(1): 74, 2023 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-37460462

RESUMO

Posttranslational modification dramatically enhances protein complexity, but the function and precise mechanism of novel lysine acylation modifications remain unknown. Chemoresistance remains a daunting challenge to successful treatment. We found that lysine butyrylation (Kbu) is specifically upregulated in chemoresistant tumor cells and tissues. By integrating butyrylome profiling and gain/loss-of-function experiments, lysine 754 in HSP90 (HSP90 K754) was identified as a substrate for Kbu. Kbu modification leads to overexpression of HSP90 in esophageal squamous cell carcinoma (ESCC) and its further increase in relapse samples. Upregulation of HSP90 contributes to 5-FU resistance and can predict poor prognosis in cancer patients. Mechanistically, HSP90 K754 is regulated by the cooperation of KAT8 and HDAC11 as the writer and eraser, respectively; SDCBP increases the Kbu level and stability of HSP90 by binding competitively to HDAC11. Furthermore, SDCBP blockade with the lead compound V020-9974 can target HSP90 K754 to overcome 5-FU resistance, constituting a potential therapeutic strategy.

20.
Zhongguo Dang Dai Er Ke Za Zhi ; 14(3): 215-20, 2012 Mar.
Artigo em Zh | MEDLINE | ID: mdl-22433412

RESUMO

OBJECTIVE: To study the effects of miR-124-1 on neuronal differentiation of rat bone marrow mesenchymal stem cells (MSCs). METHODS: MSCs cells were assigned into three groups: control (uninfected and untransfected), miR-124-1+ (infected with miR-124-1), and miR-124-1- (transfected with Anti-rno-miR-124* Inhibitor). MSCs were induced by ß-mercaptoethanol (ß-ME) to differentiate into neurons. The fluorescence expressed by infected MSCs was observed under an inverted fluorescence microscope. MTT method was used to measure cell survival rate after transfection or infection. Immunocytochemistry, RT-PCR and Western blot methods were used to detect the expression of ß3 tubulin, MAP-2 and GFAP 6 days after ß-ME induction. RESULTS: The expression of miR-124-1 in the miR-124-1+ group was significantly higher 2 days after infection of lentivirus vector compared with the control group (P<0.01). In the miR-124-1- group, the cell survival rate and the miR-124-1 expression level decreased significantly 24 hrs after transfection of anti-rno-miR-124* inhibitor (P<0.01). After 6 days of ß-ME induction, the protein and mRNA expression levels of ß3 tubulin and MAP-2 in the miR-124-1+ group were much higher than the other two groups (P<0.01); while the expression levels of ß3 tubulin and MAP-2 in the miR-124-1-group were lower than the control group (P<0.01). The expression of GFAP in the three groups was weak (<1%). CONCLUSIONS: miR-124 might promote neuronal differentiation of rat MSCs.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , MicroRNAs/fisiologia , Neurônios/citologia , Animais , Feminino , Proteína Glial Fibrilar Ácida/análise , Masculino , Proteínas Associadas aos Microtúbulos/análise , Ratos , Ratos Wistar , Tubulina (Proteína)/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA